1
|
Reyes R, Rodriguez-Muñoz R, Nahmad M. Cell recruitment and the origins of Anterior-Posterior asymmetries in the Drosophila wing. PLoS One 2025; 20:e0313067. [PMID: 39752433 PMCID: PMC11698434 DOI: 10.1371/journal.pone.0313067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 10/17/2024] [Indexed: 01/06/2025] Open
Abstract
The mechanisms underlying the establishment of asymmetric structures during development remain elusive. The wing of Drosophila is asymmetric along the Anterior-Posterior (AP) axis, but the developmental origins of this asymmetry is unknown. Here, we investigate the contribution of cell recruitment, a process that drives cell fate differentiation in the Drosophila wing disc, to the asymmetric shape and pattern of the adult wing. Genetic impairment of cell recruitment in the wing disc results in a significant gain of AP symmetry, which results from a reduction of the region between longitudinal vein 5 and the wing margin (L5-M) in the adult wing. Morphometric analysis confirms that blocking of cell recruitment results in a more symmetric wing with respect to controls, suggesting a contribution of cell recruitment to the establishment of asymmetry in the adult wing. In order to verify if this phenotype is originated during the time in which cell recruitment occurs during larval development, we examined the expression of a reporter for the selector gene vestigial (vg) in the corresponding pro-vein regions of the wing disc, but our findings could not explain our findings in adult wings. However, the circularity of the Vg pattern significantly increases in recruitment-impaired wing discs, suggesting that cell recruitment may contribute to AP asymmetries in the adult wing shape by altering the roundness of the Vg pattern. We conclude that cell recruitment, a widespread mechanism that participates in growth and patterning of several developing systems, may contribute, at least partially, to the asymmetric shape of the Drosophila wing.
Collapse
Affiliation(s)
- Rosalío Reyes
- Department of Physiology, Biophysics, and Neurosciences; Center for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
- Interdisciplinary Polytechnic Unit of Biotechnology of the National Polytechnic Institute, Mexico City, Mexico
| | - Rafael Rodriguez-Muñoz
- Department of Physiology, Biophysics, and Neurosciences; Center for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Marcos Nahmad
- Department of Physiology, Biophysics, and Neurosciences; Center for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| |
Collapse
|
2
|
Liu A, O’Connell J, Wall F, Carthew RW. Scaling between cell cycle duration and wing growth is regulated by Fat-Dachsous signaling in Drosophila. eLife 2024; 12:RP91572. [PMID: 38842917 PMCID: PMC11156469 DOI: 10.7554/elife.91572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024] Open
Abstract
The atypical cadherins Fat and Dachsous (Ds) signal through the Hippo pathway to regulate growth of numerous organs, including the Drosophila wing. Here, we find that Ds-Fat signaling tunes a unique feature of cell proliferation found to control the rate of wing growth during the third instar larval phase. The duration of the cell cycle increases in direct proportion to the size of the wing, leading to linear-like growth during the third instar. Ds-Fat signaling enhances the rate at which the cell cycle lengthens with wing size, thus diminishing the rate of wing growth. We show that this results in a complex but stereotyped relative scaling of wing growth with body growth in Drosophila. Finally, we examine the dynamics of Fat and Ds protein distribution in the wing, observing graded distributions that change during growth. However, the significance of these dynamics is unclear since perturbations in expression have negligible impact on wing growth.
Collapse
Affiliation(s)
- Andrew Liu
- Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
- NSF-Simons Center for Quantitative Biology, Northwestern UniversityEvanstonUnited States
- NSF-Simons National Institute for Theory and Mathematics in BiologyChicagoUnited States
| | - Jessica O’Connell
- Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
| | - Farley Wall
- Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
| | - Richard W Carthew
- Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
- NSF-Simons Center for Quantitative Biology, Northwestern UniversityEvanstonUnited States
- NSF-Simons National Institute for Theory and Mathematics in BiologyChicagoUnited States
| |
Collapse
|
3
|
Liu A, O’Connell J, Wall F, Carthew RW. Scaling between cell cycle duration and wing growth is regulated by Fat-Dachsous signaling in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.01.551465. [PMID: 38645118 PMCID: PMC11030236 DOI: 10.1101/2023.08.01.551465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The atypical cadherins Fat and Dachsous (Ds) signal through the Hippo pathway to regulate growth of numerous organs, including the Drosophila wing. Here, we find that Ds-Fat signaling tunes a unique feature of cell proliferation found to control the rate of wing growth during the third instar larval phase. The duration of the cell cycle increases in direct proportion to the size of the wing, leading to linear-like growth during the third instar. Ds-Fat signaling enhances the rate at which the cell cycle lengthens with wing size, thus diminishing the rate of wing growth. We show that this results in a complex but stereotyped relative scaling of wing growth with body growth in Drosophila. Finally, we examine the dynamics of Fat and Ds protein distribution in the wing, observing graded distributions that change during growth. However, the significance of these dynamics is unclear since perturbations in expression have negligible impact on wing growth.
Collapse
Affiliation(s)
- Andrew Liu
- Department of Molecular Biosciences, Northwestern University, Evanston IL
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston IL
| | - Jessica O’Connell
- Department of Molecular Biosciences, Northwestern University, Evanston IL
| | - Farley Wall
- Department of Molecular Biosciences, Northwestern University, Evanston IL
| | - Richard W. Carthew
- Department of Molecular Biosciences, Northwestern University, Evanston IL
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston IL
| |
Collapse
|
4
|
Brown HE, Weasner BP, Weasner BM, Kumar JP. Polycomb safeguards imaginal disc specification through control of the Vestigial-Scalloped complex. Development 2023; 150:dev201872. [PMID: 37702007 PMCID: PMC10560572 DOI: 10.1242/dev.201872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 09/04/2023] [Indexed: 09/14/2023]
Abstract
A fundamental goal of developmental biology is to understand how cell and tissue fates are specified. The imaginal discs of Drosophila are excellent model systems for addressing this paradigm as their fate can be redirected when discs regenerate after injury or when key selector genes are misregulated. Here, we show that when Polycomb expression is reduced, the wing selector gene vestigial is ectopically activated. This leads to the inappropriate formation of the Vestigial-Scalloped complex, which forces the eye to transform into a wing. We further demonstrate that disrupting this complex does not simply block wing formation or restore eye development. Instead, immunohistochemistry and high-throughput genomic analysis show that the eye-antennal disc unexpectedly undergoes hyperplastic growth with multiple domains being organized into other imaginal discs and tissues. These findings provide insight into the complex developmental landscape that tissues must navigate before adopting their final fate.
Collapse
Affiliation(s)
- Haley E. Brown
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | | - Bonnie M. Weasner
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Justin P. Kumar
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
5
|
Matsuda S, Affolter M. Is Drosophila Dpp/BMP morphogen spreading required for wing patterning and growth? Bioessays 2023; 45:e2200218. [PMID: 37452394 DOI: 10.1002/bies.202200218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 06/02/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023]
Abstract
Secreted signaling molecules act as morphogens to control patterning and growth in many developing tissues. Since locally produced morphogens spread to form a concentration gradient in the surrounding tissue, spreading is generally thought to be the key step in the non-autonomous actions. Here, we review recent advances in tool development to investigate morphogen function using the role of decapentaplegic (Dpp)/bone morphogenetic protein (BMP)-type ligand in the Drosophila wing disc as an example. By applying protein binder tools to distinguish between the roles of Dpp spreading and local Dpp signaling, we found that Dpp signaling in the source cells is important for wing patterning and growth but Dpp spreading from this source cells is not as strictly required as previously thought. Given recent studies showing unexpected requirements of long-range action of different morphogens, manipulating endogenous morphogen gradients by synthetic protein binder tools could shed more light on how morphogens act in developing tissues.
Collapse
Affiliation(s)
- Shinya Matsuda
- Growth & Development, Biozentrum, University of Basel, Basel, Switzerland
| | - Markus Affolter
- Growth & Development, Biozentrum, University of Basel, Basel, Switzerland
| |
Collapse
|
6
|
Yamashita T, Ohde T, Nakamura T, Ishimaru Y, Watanabe T, Tomonari S, Nakamura Y, Noji S, Mito T. Involvement of the scalloped gene in morphogenesis of the wing margin via regulating cell growth in a hemimetabolous insect Gryllus bimaculatus. Dev Growth Differ 2023; 65:348-359. [PMID: 37310211 DOI: 10.1111/dgd.12869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 06/14/2023]
Abstract
The acquisition of wings was a key event in insect evolution. As hemimetabolous insects were the first group to acquire functional wings, establishing the mechanisms of wing formation in this group could provide useful insights into their evolution. In this study, we aimed to elucidate the expression and function of the gene scalloped (sd), which is involved in wing formation in Drosophila melanogaster, and in Gryllus bimaculatus mainly during postembryonic development. Expression analysis showed that sd is expressed in the tergal edge, legs, antennae, labrum, and cerci during embryogenesis and in the distal margin of the wing pads from at least the sixth instar in the mid to late stages. Because sd knockout caused early lethality, nymphal RNA interference experiments were performed. Malformations were observed in the wings, ovipositor, and antennae. By analyzing the effects on wing morphology, it was revealed that sd is mainly involved in the formation of the margin, possibly through the regulation of cell proliferation. In conclusion, sd might regulate the local growth of wing pads and influence wing margin morphology in Gryllus.
Collapse
Grants
- 17H03945 Ministry of Education, Culture, Sports, Science and Technology
- 19H02970 Ministry of Education, Culture, Sports, Science and Technology
- 19K06691 Ministry of Education, Culture, Sports, Science and Technology
- 20K21436 Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Takahisa Yamashita
- Graduate School of Advanced Technology and Science, Tokushima University, Tokushima, Japan
| | - Takahiro Ohde
- Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Taro Nakamura
- Division of Evolutionary Developmental Biology, National Institute for Basic Biology, Okazaki, Japan
| | - Yoshiyasu Ishimaru
- Graduate School of Sciences and Technology for Innovation, Tokushima University, Tokushima, Japan
| | - Takahito Watanabe
- Bio-Innovation Research Center, Tokushima University, Tokushima, Japan
| | - Sayuri Tomonari
- Technical Support Department, Tokushima University, Tokushima, Japan
| | - Yuki Nakamura
- Graduate School of Advanced Technology and Science, Tokushima University, Tokushima, Japan
| | - Sumihare Noji
- Bio-Innovation Research Center, Tokushima University, Tokushima, Japan
| | - Taro Mito
- Bio-Innovation Research Center, Tokushima University, Tokushima, Japan
| |
Collapse
|
7
|
Farfán-Pira KJ, Martínez-Cuevas TI, Evans TA, Nahmad M. A cis-regulatory sequence of the selector gene vestigial drives the evolution of wing scaling in Drosophila species. J Exp Biol 2023; 226:jeb244692. [PMID: 37078652 PMCID: PMC10234621 DOI: 10.1242/jeb.244692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 04/13/2023] [Indexed: 04/21/2023]
Abstract
Scaling between specific organs and overall body size has long fascinated biologists, being a primary mechanism by which organ shapes evolve. Yet, the genetic mechanisms that underlie the evolution of scaling relationships remain elusive. Here, we compared wing and fore tibia lengths (the latter as a proxy of body size) in Drosophila melanogaster, Drosophila simulans, Drosophila ananassae and Drosophila virilis, and show that the first three of these species have roughly a similar wing-to-tibia scaling behavior. In contrast, D. virilis exhibits much smaller wings relative to their body size compared with the other species and this is reflected in the intercept of the wing-to-tibia allometry. We then asked whether the evolution of this relationship could be explained by changes in a specific cis-regulatory region or enhancer that drives expression of the wing selector gene, vestigial (vg), whose function is broadly conserved in insects and contributes to wing size. To test this hypothesis directly, we used CRISPR/Cas9 to replace the DNA sequence of the predicted Quadrant Enhancer (vgQE) from D. virilis for the corresponding vgQE sequence in the genome of D. melanogaster. Strikingly, we discovered that D. melanogaster flies carrying the D. virilis vgQE sequence have wings that are significantly smaller with respect to controls, partially shifting the intercept of the wing-to-tibia scaling relationship towards that observed in D. virilis. We conclude that a single cis-regulatory element in D. virilis contributes to constraining wing size in this species, supporting the hypothesis that scaling could evolve through genetic variations in cis-regulatory elements.
Collapse
Affiliation(s)
- Keity J. Farfán-Pira
- Department of Physiology, Biophysics and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City 07360, Mexico
| | - Teresa I. Martínez-Cuevas
- Department of Physiology, Biophysics and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City 07360, Mexico
| | - Timothy A. Evans
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | - Marcos Nahmad
- Department of Physiology, Biophysics and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City 07360, Mexico
| |
Collapse
|
8
|
Flores-Flores M, Muñoz-Nava LM, Rodríguez-Muñoz R, Zartman J, Nahmad M. Vestigial-dependent induction contributes to robust patterning but is not essential for wing-fate recruitment in Drosophila. Biol Open 2023; 12:bio059908. [PMID: 37199309 PMCID: PMC10214856 DOI: 10.1242/bio.059908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 03/16/2023] [Indexed: 05/19/2023] Open
Abstract
Cell recruitment is a process by which a differentiated cell induces neighboring cells to adopt its same cell fate. In Drosophila, cells expressing the protein encoded by the wing selector gene, vestigial (vg), drive a feed-forward recruitment signal that expands the Vg pattern as a wave front. However, previous studies on Vg pattern formation do not reveal these dynamics. Here, we use live imaging to show that multiple cells at the periphery of the wing disc simultaneously activate a fluorescent reporter of the recruitment signal, suggesting that cells may be recruited without the need for their contact neighbors be recruited in advance. In support of this observation, when Vg expression is inhibited either at the dorsal-ventral boundary or away from it, the activation of the recruitment signal still occurs at a distance, suggesting that Vg expression is not absolutely required to send or propagate the recruitment signal. However, the strength and extent of the recruitment signal is clearly compromised. We conclude that a feed-forward, contact-dependent cell recruitment process is not essential for Vg patterning, but it is necessary for robustness. Overall, our findings reveal a previously unidentified role of cell recruitment as a robustness-conferring cell differentiation mechanism.
Collapse
Affiliation(s)
- Marycruz Flores-Flores
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City 07360, Mexico
- Department of Chemical and Biomolecular Engineering, Notre Dame University, Notre Dame, IN 46556, USA
| | - Luis Manuel Muñoz-Nava
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City 07360, Mexico
| | - Rafael Rodríguez-Muñoz
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City 07360, Mexico
| | - Jeremiah Zartman
- Department of Chemical and Biomolecular Engineering, Notre Dame University, Notre Dame, IN 46556, USA
| | - Marcos Nahmad
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City 07360, Mexico
| |
Collapse
|
9
|
Brown HE, Weasner BP, Weasner BM, Kumar JP. Polycomb safeguards imaginal disc specification through control of the Vestigial-Scalloped complex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.11.536444. [PMID: 37090526 PMCID: PMC10120697 DOI: 10.1101/2023.04.11.536444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
A fundamental goal of developmental biology is to understand how cell and tissue fates are specified. The imaginal discs of Drosophila are excellent model systems for addressing this paradigm as their fate can be redirected when discs regenerate after injury or when key selector genes are mis-regulated. Here, we show that when Polycomb expression is reduced, the wing selector gene vestigial is ectopically activated. This leads to the inappropriate formation of the Vestigial-Scalloped complex which forces the eye to transform into a wing. We further demonstrate that disrupting this complex does not simply block wing formation or restore eye development. Instead, immunohistochemistry and high throughput genomic analysis show that the eye-antennal disc unexpectedly undergoes hyperplastic growth with multiple domains being organized into other imaginal discs and tissues. These findings provide insight into the complex developmental landscape that tissues must navigate before adopting their final fate. Summary Statement Here we describe a novel mechanism by which Pc promotes an eye fate during normal development and how the eye is reprogrammed into a wing in its absence.
Collapse
Affiliation(s)
- Haley E. Brown
- Department of Biology, Indiana University, Bloomington, IN 47405 USA
| | | | - Bonnie M. Weasner
- Department of Biology, Indiana University, Bloomington, IN 47405 USA
| | - Justin P. Kumar
- Department of Biology, Indiana University, Bloomington, IN 47405 USA
| |
Collapse
|
10
|
Growth anisotropy of the extracellular matrix shapes a developing organ. Nat Commun 2023; 14:1220. [PMID: 36869053 PMCID: PMC9984492 DOI: 10.1038/s41467-023-36739-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 02/15/2023] [Indexed: 03/05/2023] Open
Abstract
Final organ size and shape result from volume expansion by growth and shape changes by contractility. Complex morphologies can also arise from differences in growth rate between tissues. We address here how differential growth guides the morphogenesis of the growing Drosophila wing imaginal disc. We report that 3D morphology results from elastic deformation due to differential growth anisotropy between the epithelial cell layer and its enveloping extracellular matrix (ECM). While the tissue layer grows in plane, growth of the bottom ECM occurs in 3D and is reduced in magnitude, thereby causing geometric frustration and tissue bending. The elasticity, growth anisotropy and morphogenesis of the organ are fully captured by a mechanical bilayer model. Moreover, differential expression of the Matrix metalloproteinase MMP2 controls growth anisotropy of the ECM envelope. This study shows that the ECM is a controllable mechanical constraint whose intrinsic growth anisotropy directs tissue morphogenesis in a developing organ.
Collapse
|
11
|
Diaz-Torres E, Muñoz-Nava LM, Nahmad M. Coupling cell proliferation rates to the duration of recruitment controls final size of the Drosophila wing. Proc Biol Sci 2022; 289:20221167. [PMID: 36476003 PMCID: PMC9554725 DOI: 10.1098/rspb.2022.1167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/15/2022] [Indexed: 12/14/2022] Open
Abstract
Organ growth driven by cell proliferation is an exponential process. As a result, even small variations in proliferation rates, when integrated over a relatively long developmental time, will lead to large differences in size. How organs robustly control their final size despite perturbations in cell proliferation rates throughout development is a long-standing question in biology. Using a mathematical model, we show that in the developing wing of the fruit fly, Drosophila melanogaster, variations in proliferation rates of wing-committed cells are inversely proportional to the duration of cell recruitment, a differentiation process in which a population of undifferentiated cells adopt the wing fate by expressing the selector gene, vestigial. A time-course experiment shows that vestigial-expressing cells increase exponentially while recruitment takes place, but slows down when recruitable cells start to vanish, suggesting that undifferentiated cells may be driving proliferation of wing-committed cells. When this observation is incorporated in our model, we show that the duration of cell recruitment robustly determines a final wing size even when cell proliferation rates of wing-committed cells are perturbed. Finally, we show that this control mechanism fails when perturbations in proliferation rates affect both wing-committed and recruitable cells, providing an experimentally testable hypothesis of our model.
Collapse
Affiliation(s)
- Elizabeth Diaz-Torres
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Av. Instituto Politecnico Nacional 2508, Colonia San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Luis Manuel Muñoz-Nava
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Av. Instituto Politecnico Nacional 2508, Colonia San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Marcos Nahmad
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Av. Instituto Politecnico Nacional 2508, Colonia San Pedro Zacatenco, Mexico City 07360, Mexico
| |
Collapse
|
12
|
Farfán-Pira KJ, Martínez-Cuevas TI, Reyes R, Evans TA, Nahmad M. The vestigial Quadrant Enhancer is dispensable for pattern formation and development of the Drosophila wing. MICROPUBLICATION BIOLOGY 2022; 2022:10.17912/micropub.biology.000585. [PMID: 35783575 PMCID: PMC9242444 DOI: 10.17912/micropub.biology.000585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 11/06/2022]
Abstract
In Drosophila , the pattern of the wing selector gene, vestigial ( vg ), is established by at least two enhancers: the Boundary Enhancer, which drives expression along the disc's Dorsal-Ventral boundary; and the Quadrant Enhancer (QE) that patterns the rest of the wing pouch. Using CRISPR/Cas9 editing, we deleted DNA fragments around the reported QE sequence and found that the full Vg pattern is formed. Furthermore, adult wings arising from these gene-edited animals are normal in shape and pattern, but slightly smaller in size, although this reduction is not wing-specific in males. We suggest that other enhancers act redundantly to establish the vg pattern and rescue wing development.
Collapse
Affiliation(s)
- Keity J Farfán-Pira
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN)
| | - Teresa I Martínez-Cuevas
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN)
| | - Rosalio Reyes
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN)
| | | | - Marcos Nahmad
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN)
,
Correspondence to: Marcos Nahmad (
)
| |
Collapse
|
13
|
Hecht S, Perez-Mockus G, Schienstock D, Recasens-Alvarez C, Merino-Aceituno S, Smith M, Salbreux G, Degond P, Vincent JP. Mechanical constraints to cell-cycle progression in a pseudostratified epithelium. Curr Biol 2022; 32:2076-2083.e2. [PMID: 35338851 PMCID: PMC7615048 DOI: 10.1016/j.cub.2022.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/14/2021] [Accepted: 03/01/2022] [Indexed: 02/07/2023]
Abstract
As organs and tissues approach their normal size during development or regeneration, growth slows down, and cell proliferation progressively comes to a halt. Among the various processes suggested to contribute to growth termination,1-10 mechanical feedback, perhaps via adherens junctions, has been suggested to play a role.11-14 However, since adherens junctions are only present in a narrow plane of the subapical region, other structures are likely needed to sense mechanical stresses along the apical-basal (A-B) axis, especially in a thick pseudostratified epithelium. This could be achieved by nuclei, which have been implicated in mechanotransduction in tissue culture.15 In addition, mechanical constraints imposed by nuclear crowding and spatial confinement could affect interkinetic nuclear migration (IKNM),16 which allows G2 nuclei to reach the apical surface, where they normally undergo mitosis.17-25 To explore how mechanical constraints affect IKNM, we devised an individual-based model that treats nuclei as deformable objects constrained by the cell cortex and the presence of other nuclei. The model predicts changes in the proportion of cell-cycle phases during growth, which we validate with the cell-cycle phase reporter FUCCI (Fluorescent Ubiquitination-based Cell Cycle Indicator).26 However, this model does not preclude indefinite growth, leading us to postulate that nuclei must migrate basally to access a putative basal signal required for S phase entry. With this refinement, our updated model accounts for the observed progressive slowing down of growth and explains how pseudostratified epithelia reach a stereotypical thickness upon completion of growth.
Collapse
Affiliation(s)
- Sophie Hecht
- The Francis Crick Institute, London NW1 1AT, UK; Imperial College London, Department of Mathematics, London SW7 2AZ, UK
| | | | | | | | - Sara Merino-Aceituno
- University of Vienna, Faculty of Mathematics, Oskar-Morgenstern-Platz 1, Wien 1090, Austria; University of Sussex, Department of Mathematics, Falmer BN1 9RH, UK
| | - Matt Smith
- The Francis Crick Institute, London NW1 1AT, UK
| | | | - Pierre Degond
- Imperial College London, Department of Mathematics, London SW7 2AZ, UK.
| | | |
Collapse
|
14
|
Ohde T, Mito T, Niimi T. A hemimetabolous wing development suggests the wing origin from lateral tergum of a wingless ancestor. Nat Commun 2022; 13:979. [PMID: 35190538 PMCID: PMC8861169 DOI: 10.1038/s41467-022-28624-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 01/24/2022] [Indexed: 11/26/2022] Open
Abstract
The origin and evolution of the novel insect wing remain enigmatic after a century-long discussion. The mechanism of wing development in hemimetabolous insects, in which the first functional wings evolved, is key to understand where and how insect wings evolutionarily originate. This study explored the developmental origin and the postembryonic dramatic growth of wings in the cricket Gryllus bimaculatus. We find that the lateral tergal margin, which is homologous between apterygote and pterygote insects, comprises a growth organizer to expand the body wall to form adult wing blades in Gryllus. We also find that Wnt, Fat-Dachsous, and Hippo pathways are involved in the disproportional growth of Gryllus wings. These data provide insights into where and how insect wings originate. Wings evolved from the pre-existing lateral terga of a wingless insect ancestor, and the reactivation or redeployment of Wnt/Fat-Dachsous/Hippo-mediated feed-forward circuit might have expanded the lateral terga. Here, the authors investigate wing development in cricket and find support for evolution of the novel insect wing from the pre-existing dorsal body wall of a wingless ancestor by activation of an evolutionarily conserved growth mechanism.
Collapse
|
15
|
Abstract
The Drosophila wing imaginal disc is a tissue of undifferentiated cells that are precursors of the wing and most of the notum of the adult fly. The wing disc first forms during embryogenesis from a cluster of ∼30 cells located in the second thoracic segment, which invaginate to form a sac-like structure. They undergo extensive proliferation during larval stages to form a mature larval wing disc of ∼35,000 cells. During this time, distinct cell fates are assigned to different regions, and the wing disc develops a complex morphology. Finally, during pupal stages the wing disc undergoes morphogenetic processes and then differentiates to form the adult wing and notum. While the bulk of the wing disc comprises epithelial cells, it also includes neurons and glia, and is associated with tracheal cells and muscle precursor cells. The relative simplicity and accessibility of the wing disc, combined with the wealth of genetic tools available in Drosophila, have combined to make it a premier system for identifying genes and deciphering systems that play crucial roles in animal development. Studies in wing imaginal discs have made key contributions to many areas of biology, including tissue patterning, signal transduction, growth control, regeneration, planar cell polarity, morphogenesis, and tissue mechanics.
Collapse
Affiliation(s)
- Bipin Kumar Tripathi
- Department of Molecular Biology and Biochemistry, Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Kenneth D Irvine
- Department of Molecular Biology and Biochemistry, Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
16
|
Matsuda S, Schaefer JV, Mii Y, Hori Y, Bieli D, Taira M, Plückthun A, Affolter M. Asymmetric requirement of Dpp/BMP morphogen dispersal in the Drosophila wing disc. Nat Commun 2021; 12:6435. [PMID: 34750371 PMCID: PMC8576045 DOI: 10.1038/s41467-021-26726-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 10/20/2021] [Indexed: 11/26/2022] Open
Abstract
How morphogen gradients control patterning and growth in developing tissues remains largely unknown due to lack of tools manipulating morphogen gradients. Here, we generate two membrane-tethered protein binders that manipulate different aspects of Decapentaplegic (Dpp), a morphogen required for overall patterning and growth of the Drosophila wing. One is "HA trap" based on a single-chain variable fragment (scFv) against the HA tag that traps HA-Dpp to mainly block its dispersal, the other is "Dpp trap" based on a Designed Ankyrin Repeat Protein (DARPin) against Dpp that traps Dpp to block both its dispersal and signaling. Using these tools, we found that, while posterior patterning and growth require Dpp dispersal, anterior patterning and growth largely proceed without Dpp dispersal. We show that dpp transcriptional refinement from an initially uniform to a localized expression and persistent signaling in transient dpp source cells render the anterior compartment robust against the absence of Dpp dispersal. Furthermore, despite a critical requirement of dpp for the overall wing growth, neither Dpp dispersal nor direct signaling is critical for lateral wing growth after wing pouch specification. These results challenge the long-standing dogma that Dpp dispersal is strictly required to control and coordinate overall wing patterning and growth.
Collapse
Affiliation(s)
| | - Jonas V Schaefer
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Yusuke Mii
- National Institute for Basic Biology and Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- JST PRESTO, Kawaguchi, Saitama, Japan
| | - Yutaro Hori
- Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | | | - Masanori Taira
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- Department of Biological Sciences, Faculty of Science and Engineering, Chuo University, Tokyo, Japan
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
17
|
Magny EG, Platero AI, Bishop SA, Pueyo JI, Aguilar-Hidalgo D, Couso JP. Pegasus, a small extracellular peptide enhancing short-range diffusion of Wingless. Nat Commun 2021; 12:5660. [PMID: 34580289 PMCID: PMC8476528 DOI: 10.1038/s41467-021-25785-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 07/08/2021] [Indexed: 11/09/2022] Open
Abstract
Small Open Reading Frames (smORFs) coding for peptides of less than 100 amino-acids are an enigmatic and pervasive gene class, found in the tens of thousands in metazoan genomes. Here we reveal a short 80 amino-acid peptide (Pegasus) which enhances Wingless/Wnt1 protein short-range diffusion and signalling. During Drosophila wing development, Wingless has sequential functions, including late induction of proneural gene expression and wing margin development. Pegasus mutants produce wing margin defects and proneural expression loss similar to those of Wingless. Pegasus is secreted, and co-localizes and co-immunoprecipitates with Wingless, suggesting their physical interaction. Finally, measurements of fixed and in-vivo Wingless gradients support that Pegasus increases Wingless diffusion in order to enhance its signalling. Our results unveil a new element in Wingless signalling and clarify the patterning role of Wingless diffusion, while corroborating the link between small open reading frame peptides, and regulation of known proteins with membrane-related functions.
Collapse
Affiliation(s)
- Emile G Magny
- Centro Andaluz de Biologia del Desarrollo, CSIC-Universidad Pablo de Olavide, Sevilla, Spain
| | - Ana Isabel Platero
- Centro Andaluz de Biologia del Desarrollo, CSIC-Universidad Pablo de Olavide, Sevilla, Spain
| | - Sarah A Bishop
- Centro Andaluz de Biologia del Desarrollo, CSIC-Universidad Pablo de Olavide, Sevilla, Spain.,Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Jose I Pueyo
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Daniel Aguilar-Hidalgo
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Juan Pablo Couso
- Centro Andaluz de Biologia del Desarrollo, CSIC-Universidad Pablo de Olavide, Sevilla, Spain.
| |
Collapse
|
18
|
Jiang TX, Li A, Lin CM, Chiu C, Cho JH, Reid B, Zhao M, Chow RH, Widelitz RB, Chuong CM. Global feather orientations changed by electric current. iScience 2021; 24:102671. [PMID: 34179734 PMCID: PMC8214094 DOI: 10.1016/j.isci.2021.102671] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/18/2021] [Accepted: 05/27/2021] [Indexed: 12/17/2022] Open
Abstract
During chicken skin development, each feather bud exhibits its own polarity, but a population of buds organizes with a collective global orientation. We used embryonic dorsal skin, with buds aligned parallel to the rostral-caudal body axis, to explore whether exogenous electric fields affect feather polarity. Interestingly, brief exogenous current exposure prior to visible bud formation later altered bud orientations. Applying electric pulses perpendicular to the body rostral-caudal axis realigned bud growth in a collective swirl, resembling an electric field pointing toward the anode. Perturbed buds show normal molecular expression and morphogenesis except for their altered orientation. Epithelial-mesenchymal recombination demonstrates the effects of exogenous electric fields are mediated through the epithelium. Small-molecule channel inhibitor screens show Ca2+ channels and PI3 Kinase are involved in controlling feather bud polarity. This work reveals the importance of bioelectricity in organ development and regeneration and provides an explant culture platform for experimentation.
Collapse
Affiliation(s)
- Ting-Xin Jiang
- Department of Pathology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, Los Ángeles, CA 90033, USA
| | - Ang Li
- Department of Pathology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, Los Ángeles, CA 90033, USA
| | - Chih-Min Lin
- Department of Pathology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, Los Ángeles, CA 90033, USA
| | - Cathleen Chiu
- Department of Pathology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, Los Ángeles, CA 90033, USA
| | - Jung-Hwa Cho
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Brian Reid
- Department of Ophthalmology & Vision Science, and Department of Dermatology, University of California, Davis, Sacramento, CA 95816, USA
| | - Min Zhao
- Department of Ophthalmology & Vision Science, and Department of Dermatology, University of California, Davis, Sacramento, CA 95816, USA
| | - Robert H. Chow
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Randall Bruce Widelitz
- Department of Pathology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, Los Ángeles, CA 90033, USA
| | - Cheng-Ming Chuong
- Department of Pathology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, Los Ángeles, CA 90033, USA
| |
Collapse
|
19
|
Lee J, Ng KGL, Dombek KM, Eom DS, Kwon YV. Tumors overcome the action of the wasting factor ImpL2 by locally elevating Wnt/Wingless. Proc Natl Acad Sci U S A 2021; 118:e2020120118. [PMID: 34078667 PMCID: PMC8201939 DOI: 10.1073/pnas.2020120118] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Tumors often secrete wasting factors associated with atrophy and the degeneration of host tissues. If tumors were to be affected by the wasting factors, mechanisms allowing tumors to evade the adverse effects of the wasting factors must exist, and impairing such mechanisms may attenuate tumors. We use Drosophila midgut tumor models to show that tumors up-regulate Wingless (Wg) to oppose the growth-impeding effects caused by the wasting factor, ImpL2 (insulin-like growth factor binding protein [IGFBP]-related protein). Growth of Yorkie (Yki)-induced tumors is dependent on Wg while either elimination of ImpL2 or elevation of insulin/insulin-like growth factor signaling in tumors revokes this dependency. Notably, Wg augmentation could be a general mechanism for supporting the growth of tumors with elevated ImpL2 and exploited to attenuate muscle degeneration during wasting. Our study elucidates the mechanism by which tumors negate the action of ImpL2 to uphold their growth during cachexia-like wasting and implies that targeting the Wnt/Wg pathway might be an efficient treatment strategy for cancers with elevated IGFBPs.
Collapse
Affiliation(s)
- Jiae Lee
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA 98195
| | - Katelyn G-L Ng
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA 98195
| | - Kenneth M Dombek
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA 98195
| | - Dae Seok Eom
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, CA 92697
| | - Young V Kwon
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA 98195;
| |
Collapse
|
20
|
Estella C, Baonza A. Cell proliferation control by Notch signalling during imaginal discs development in Drosophila. AIMS GENETICS 2021. [DOI: 10.3934/genet.2015.1.70] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
AbstractThe Notch signalling pathway is evolutionary conserved and participates in numerous developmental processes, including the control of cell proliferation. However, Notch signalling can promote or restrain cell division depending on the developmental context, as has been observed in human cancer where Notch can function as a tumor suppressor or an oncogene. Thus, the outcome of Notch signalling can be influenced by the cross-talk between Notch and other signalling pathways. The use of model organisms such as Drosophila has been proven to be very valuable to understand the developmental role of the Notch pathway in different tissues and its relationship with other signalling pathways during cell proliferation control. Here we review recent studies in Drosophila that shed light in the developmental control of cell proliferation by the Notch pathway in different contexts such as the eye, wing and leg imaginal discs. We also discuss the autonomous and non-autonomous effects of the Notch pathway on cell proliferation and its interactions with different signalling pathways.
Collapse
Affiliation(s)
- Carlos Estella
- Departamento de Biología Molecular and Centro de Biología Molecular SeveroOchoa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Antonio Baonza
- Centro de Biología Molecular Severo Ochoa (CSIC/UAM) c/Nicolás Cabrera 1, 28049, Madrid, Spain
| |
Collapse
|
21
|
Zecca M, Struhl G. A unified mechanism for the control of Drosophila wing growth by the morphogens Decapentaplegic and Wingless. PLoS Biol 2021; 19:e3001111. [PMID: 33657096 PMCID: PMC8148325 DOI: 10.1371/journal.pbio.3001111] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 05/25/2021] [Accepted: 01/22/2021] [Indexed: 12/31/2022] Open
Abstract
Development of the Drosophila wing-a paradigm of organ development-is governed by 2 morphogens, Decapentaplegic (Dpp, a BMP) and Wingless (Wg, a Wnt). Both proteins are produced by defined subpopulations of cells and spread outwards, forming gradients that control gene expression and cell pattern as a function of concentration. They also control growth, but how is unknown. Most studies have focused on Dpp and yielded disparate models in which cells throughout the wing grow at similar rates in response to the grade or temporal change in Dpp concentration or to the different amounts of Dpp "equalized" by molecular or mechanical feedbacks. In contrast, a model for Wg posits that growth is governed by a progressive expansion in morphogen range, via a mechanism in which a minimum threshold of Wg sustains the growth of cells within the wing and recruits surrounding "pre-wing" cells to grow and enter the wing. This mechanism depends on the capacity of Wg to fuel the autoregulation of vestigial (vg)-the selector gene that specifies the wing state-both to sustain vg expression in wing cells and by a feed-forward (FF) circuit of Fat (Ft)/Dachsous (Ds) protocadherin signaling to induce vg expression in neighboring pre-wing cells. Here, we have subjected Dpp to the same experimental tests used to elucidate the Wg model and find that it behaves indistinguishably. Hence, we posit that both morphogens act together, via a common mechanism, to control wing growth as a function of morphogen range.
Collapse
Affiliation(s)
- Myriam Zecca
- Department of Genetics and Development, Columbia University, New York, New York, United States of America
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
| | - Gary Struhl
- Department of Genetics and Development, Columbia University, New York, New York, United States of America
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
| |
Collapse
|
22
|
Control of Drosophila wing size by morphogen range and hormonal gating. Proc Natl Acad Sci U S A 2020; 117:31935-31944. [PMID: 33257577 DOI: 10.1073/pnas.2018196117] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The stereotyped dimensions of animal bodies and their component parts result from tight constraints on growth. Yet, the mechanisms that stop growth when organs reach the right size are unknown. Growth of the Drosophila wing-a classic paradigm-is governed by two morphogens, Decapentaplegic (Dpp, a BMP) and Wingless (Wg, a Wnt). Wing growth during larval life ceases when the primordium attains full size, concomitant with the larval-to-pupal molt orchestrated by the steroid hormone ecdysone. Here, we block the molt by genetically dampening ecdysone production, creating an experimental paradigm in which the wing stops growing at the correct size while the larva continues to feed and gain body mass. Under these conditions, we show that wing growth is limited by the ranges of Dpp and Wg, and by ecdysone, which regulates the cellular response to their signaling activities. Further, we present evidence that growth terminates because of the loss of two distinct modes of morphogen action: 1) maintenance of growth within the wing proper and 2) induced growth of surrounding "pre-wing" cells and their recruitment into the wing. Our results provide a precedent for the control of organ size by morphogen range and the hormonal gating of morphogen action.
Collapse
|
23
|
Lim HY, Bao H, Liu Y, Wang W. Select Septate Junction Proteins Direct ROS-Mediated Paracrine Regulation of Drosophila Cardiac Function. Cell Rep 2020; 28:1455-1470.e4. [PMID: 31390561 DOI: 10.1016/j.celrep.2019.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 04/18/2019] [Accepted: 06/27/2019] [Indexed: 12/27/2022] Open
Abstract
Septate junction (SJ) complex proteins act in unison to provide a paracellular barrier and maintain structural integrity. Here, we identify a non-barrier role of two individual SJ proteins, Coracle (Cora) and Kune-kune (Kune). Reactive oxygen species (ROS)-p38 MAPK signaling in non-myocytic pericardial cells (PCs) is important for maintaining normal cardiac physiology in Drosophila. However, the underlying mechanisms remain unknown. We find that in PCs, Cora and Kune are altered in abundance in response to manipulations of ROS-p38 signaling. Genetic analyses establish Cora and Kune as key effectors of ROS-p38 signaling in PCs on proper heart function. We further determine that Cora regulates normal Kune levels in PCs, which in turn modulates normal Kune levels in the cardiomyocytes essential for proper heart function. Our results thereby reveal select SJ proteins Cora and Kune as signaling mediators of the PC-derived ROS regulation of cardiac physiology.
Collapse
Affiliation(s)
- Hui-Ying Lim
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA.
| | - Hong Bao
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Ying Liu
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Weidong Wang
- Department of Medicine, Section of Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
24
|
Gawne R, McKenna KZ, Levin M. Competitive and Coordinative Interactions between Body Parts Produce Adaptive Developmental Outcomes. Bioessays 2020; 42:e1900245. [DOI: 10.1002/bies.201900245] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Indexed: 01/25/2023]
Affiliation(s)
- Richard Gawne
- Allen Discovery Center at Tufts University Medford MA 02155
| | - Kenneth Z. McKenna
- Division of Biological SciencesSection of Cellular and Developmental BiologyUniversity of California San Diego La Jolla CA 92093
| | - Michael Levin
- Allen Discovery Center at Tufts University Medford MA 02155
| |
Collapse
|
25
|
Muñoz-Nava LM, Alvarez HA, Flores-Flores M, Chara O, Nahmad M. A dynamic cell recruitment process drives growth of the Drosophila wing by overscaling the vestigial expression pattern. Dev Biol 2020; 462:141-151. [PMID: 32197891 DOI: 10.1016/j.ydbio.2020.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/10/2020] [Accepted: 03/11/2020] [Indexed: 10/24/2022]
Abstract
Organs mainly attain their size by cell growth and proliferation, but sometimes also grow through recruitment of undifferentiated cells. Here we investigate the participation of cell recruitment in establishing the pattern of Vestigial (Vg), the product of the wing selector gene in Drosophila. We find that the Vg pattern overscales along the dorsal-ventral (DV) axis of the wing imaginal disc, i.e., it expands faster than the DV length of the pouch. The overscaling of the Vg pattern cannot be explained by differential proliferation, apoptosis, or oriented-cell divisions, but can be recapitulated by a mathematical model that explicitly considers cell recruitment. When impairing cell recruitment genetically, we find that the Vg pattern almost perfectly scales and adult wings are approximately 20% smaller. Conversely, impairing cell proliferation results in very small wings, suggesting that cell recruitment and cell proliferation additively contribute to organ growth in this system. Furthermore, using fluorescent reporter tools, we provide direct evidence that cell recruitment is initiated between early and mid third-instar larval development. Altogether, our work quantitatively shows when, how, and by how much cell recruitment shapes the Vg pattern and drives growth of the Drosophila wing.
Collapse
Affiliation(s)
- Luis Manuel Muñoz-Nava
- Department of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnical Institute (Cinvestav-IPN), Mexico City, 07360, Mexico
| | - Hugo Ariel Alvarez
- Systems Biology Group (SysBio), Institute of Physics of Liquids and Biological Systems (IFLYSIB), National Scientific and Technical Research Council (CONICET) and University of La Plata (UNLP), La Plata, B1900BTE, Argentina; Department of Biological Sciences, Faculty of Exact Sciences, University of La Plata (UNLP), La Plata, 1900, Buenos Aires, Argentina
| | - Marycruz Flores-Flores
- Department of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnical Institute (Cinvestav-IPN), Mexico City, 07360, Mexico
| | - Osvaldo Chara
- Systems Biology Group (SysBio), Institute of Physics of Liquids and Biological Systems (IFLYSIB), National Scientific and Technical Research Council (CONICET) and University of La Plata (UNLP), La Plata, B1900BTE, Argentina; Center for Information Services and High Performance Computing (ZIH), Technische Universität Dresden (TUD), Dresden, 01069, Germany
| | - Marcos Nahmad
- Department of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnical Institute (Cinvestav-IPN), Mexico City, 07360, Mexico.
| |
Collapse
|
26
|
Li X, Liu M, Ren X, Loncle N, Wang Q, Hemba-Waduge RUS, Yu SH, Boube M, Bourbon HMG, Ni JQ, Ji JY. The Mediator CDK8-Cyclin C complex modulates Dpp signaling in Drosophila by stimulating Mad-dependent transcription. PLoS Genet 2020; 16:e1008832. [PMID: 32463833 PMCID: PMC7282676 DOI: 10.1371/journal.pgen.1008832] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/09/2020] [Accepted: 05/05/2020] [Indexed: 11/19/2022] Open
Abstract
Dysregulation of CDK8 (Cyclin-Dependent Kinase 8) and its regulatory partner CycC (Cyclin C), two subunits of the conserved Mediator (MED) complex, have been linked to diverse human diseases such as cancer. Thus, it is essential to understand the regulatory network modulating the CDK8-CycC complex in both normal development and tumorigenesis. To identify upstream regulators or downstream effectors of CDK8, we performed a dominant modifier genetic screen in Drosophila based on the defects in vein patterning caused by specific depletion or overexpression of CDK8 or CycC in developing wing imaginal discs. We identified 26 genomic loci whose haploinsufficiency can modify these CDK8- or CycC-specific phenotypes. Further analysis of two overlapping deficiency lines and mutant alleles led us to identify genetic interactions between the CDK8-CycC pair and the components of the Decapentaplegic (Dpp, the Drosophila homolog of TGFβ, or Transforming Growth Factor-β) signaling pathway. We observed that CDK8-CycC positively regulates transcription activated by Mad (Mothers against dpp), the primary transcription factor downstream of the Dpp/TGFβ signaling pathway. CDK8 can directly interact with Mad in vitro through the linker region between the DNA-binding MH1 (Mad homology 1) domain and the carboxy terminal MH2 (Mad homology 2) transactivation domain. Besides CDK8 and CycC, further analyses of other subunits of the MED complex have revealed six additional subunits that are required for Mad-dependent transcription in the wing discs: Med12, Med13, Med15, Med23, Med24, and Med31. Furthermore, our analyses confirmed the positive roles of CDK9 and Yorkie in regulating Mad-dependent gene expression in vivo. These results suggest that CDK8 and CycC, together with a few other subunits of the MED complex, may coordinate with other transcription cofactors in regulating Mad-dependent transcription during wing development in Drosophila.
Collapse
Affiliation(s)
- Xiao Li
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, United States of America
| | - Mengmeng Liu
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, United States of America
| | - Xingjie Ren
- School of Medicine, Tsinghua University, Beijing, China
| | - Nicolas Loncle
- Centre de Biologie Intégrative, Centre de Biologie du Développement, UMR5544 du CNRS, Université de Toulouse, Toulouse, France
| | - Qun Wang
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, United States of America
| | - Rajitha-Udakara-Sampath Hemba-Waduge
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, United States of America
| | - Stephen H. Yu
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, United States of America
| | - Muriel Boube
- Centre de Biologie Intégrative, Centre de Biologie du Développement, UMR5544 du CNRS, Université de Toulouse, Toulouse, France
| | - Henri-Marc G. Bourbon
- Centre de Biologie Intégrative, Centre de Biologie du Développement, UMR5544 du CNRS, Université de Toulouse, Toulouse, France
| | - Jian-Quan Ni
- School of Medicine, Tsinghua University, Beijing, China
| | - Jun-Yuan Ji
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, United States of America
- Department of Nutrition, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
27
|
Roy SS, Ghosh S. Genes regulating wing patterning in Drosophila melanogaster show reduced expression under exposure of Daminozide, the fruit ripening retardant. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 75:103322. [PMID: 31877500 DOI: 10.1016/j.etap.2019.103322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/21/2019] [Accepted: 12/19/2019] [Indexed: 06/10/2023]
Abstract
In our previous study we demonstrated that the fruit ripening retardant Daminozide or Alar causes change in life history traits, distortion of adult wing structure, DNA damage in brain cells and mutagenic effects in fruit fly Drosophila melanogaster. As a continuation of the previous study the present work is designed to explore the metabolic modification of Daminozide following ingestion, the effects of Daminozide on the expression of genes which are pivotal for wing development and molecular interactions of Daminozide with those proteins involved in wing patterning. We demonstrated through reporter gene construct assay using X-gal staining method and transgenic Drosophila melanogaster stocks that the vestigial, wingless and decapentaplegic genes in wing imaginal disc from 3rd instar larvae exhibited reduced expression when exposed to Daminozide in compare to control larvae. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) of those genes confirmed that exposure to Daminozide reduces the transcription level of those genes. In silico approach with molecular docking study revealed Daminozide may bind and interfere with the optimal functioning of expressed wing signaling proteins.
Collapse
Affiliation(s)
- Sohini Singha Roy
- Cytogenetics & Genomics Research Unit, Department of Zoology, University of Calcutta, Taraknath Palit Sikshaprangan, 35 Ballygunge Circular Road, Kolkata, West Bengal, 700019 India.
| | - Sujay Ghosh
- Cytogenetics & Genomics Research Unit, Department of Zoology, University of Calcutta, Taraknath Palit Sikshaprangan, 35 Ballygunge Circular Road, Kolkata, West Bengal, 700019 India.
| |
Collapse
|
28
|
Gou J, Stotsky JA, Othmer HG. Growth control in the Drosophila wing disk. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1478. [PMID: 31917525 DOI: 10.1002/wsbm.1478] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/02/2019] [Accepted: 12/17/2019] [Indexed: 12/16/2022]
Abstract
The regulation of size and shape is a fundamental requirement of biological development and has been a subject of scientific study for centuries, but we still lack an understanding of how organisms know when to stop growing. Imaginal wing disks of the fruit fly Drosophila melanogaster, which are precursors of the adult wings, are an archetypal tissue for studying growth control. The growth of the disks is dependent on many inter- and intra-organ factors such as morphogens, mechanical forces, nutrient levels, and hormones that influence gene expression and cell growth. Extracellular signals are transduced into gene-control signals via complex signal transduction networks, and since cells typically receive many different signals, a mechanism for integrating the signals is needed. Our understanding of the effect of morphogens on tissue-level growth regulation via individual pathways has increased significantly in the last half century, but our understanding of how multiple biochemical and mechanical signals are integrated to determine whether or not a cell decides to divide is still rudimentary. Numerous fundamental questions are involved in understanding the decision-making process, and here we review the major biochemical and mechanical pathways involved in disk development with a view toward providing a basis for beginning to understand how multiple signals can be integrated at the cell level, and how this translates into growth control at the level of the imaginal disk. This article is categorized under: Analytical and Computational Methods > Computational Methods Biological Mechanisms > Cell Signaling Models of Systems Properties and Processes > Cellular Models.
Collapse
Affiliation(s)
- Jia Gou
- School of Mathematics, University of Minnesota, Minneapolis, Minnesota
| | - Jay A Stotsky
- School of Mathematics, University of Minnesota, Minneapolis, Minnesota
| | - Hans G Othmer
- School of Mathematics, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
29
|
Vollmer J, Casares F, Iber D. Growth and size control during development. Open Biol 2018; 7:rsob.170190. [PMID: 29142108 PMCID: PMC5717347 DOI: 10.1098/rsob.170190] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/17/2017] [Indexed: 11/30/2022] Open
Abstract
The size and shape of organs are characteristic for each species. Even when organisms develop to different sizes due to varying environmental conditions, such as nutrition, organ size follows species-specific rules of proportionality to the rest of the body, a phenomenon referred to as allometry. Therefore, for a given environment, organs stop growth at a predictable size set by the species's genotype. How do organs stop growth? How can related species give rise to organs of strikingly different size? No definitive answer has been given to date. One of the major models for the studies of growth termination is the vinegar fly Drosophila melanogaster. Therefore, this review will focus mostly on work carried out in Drosophila to try to tease apart potential mechanisms and identify routes for further investigation. One general rule, found across the animal kingdom, is that the rate of growth declines with developmental time. Therefore, answers to the problem of growth termination should explain this seemingly universal fact. In addition, growth termination is intimately related to the problems of robustness (i.e. precision) and plasticity in organ size, symmetric and asymmetric organ development, and of how the ‘target’ size depends on extrinsic, environmental factors.
Collapse
Affiliation(s)
- Jannik Vollmer
- D-BSSE, ETH Zürich, Mattenstrasse 26, 4058 Basel, Switzerland.,Swiss Institute of Bioinformatics (SIB), Mattenstrasse 26, 4058 Basel, Switzerland
| | - Fernando Casares
- CABD, CSIC-Universidad Pablo de Olavide-JA, 41013 Seville, Spain
| | - Dagmar Iber
- D-BSSE, ETH Zürich, Mattenstrasse 26, 4058 Basel, Switzerland .,Swiss Institute of Bioinformatics (SIB), Mattenstrasse 26, 4058 Basel, Switzerland
| |
Collapse
|
30
|
Worley MI, Alexander LA, Hariharan IK. CtBP impedes JNK- and Upd/STAT-driven cell fate misspecifications in regenerating Drosophila imaginal discs. eLife 2018; 7:30391. [PMID: 29372681 PMCID: PMC5823544 DOI: 10.7554/elife.30391] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 01/19/2018] [Indexed: 12/27/2022] Open
Abstract
Regeneration following tissue damage often necessitates a mechanism for cellular re-programming, so that surviving cells can give rise to all cell types originally found in the damaged tissue. This process, if unchecked, can also generate cell types that are inappropriate for a given location. We conducted a screen for genes that negatively regulate the frequency of notum-to-wing transformations following genetic ablation and regeneration of the wing pouch, from which we identified mutations in the transcriptional co-repressor C-terminal Binding Protein (CtBP). When CtBP function is reduced, ablation of the pouch can activate the JNK/AP-1 and JAK/STAT pathways in the notum to destabilize cell fates. Ectopic expression of Wingless and Dilp8 precede the formation of the ectopic pouch, which is subsequently generated by recruitment of both anterior and posterior cells near the compartment boundary. Thus, CtBP stabilizes cell fates following damage by opposing the destabilizing effects of the JNK/AP-1 and JAK/STAT pathways.
Collapse
Affiliation(s)
- Melanie I Worley
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Larissa A Alexander
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Iswar K Hariharan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
31
|
Ochoa-Espinosa A, Harmansa S, Caussinus E, Affolter M. Myosin II is not required for Drosophila tracheal branch elongation and cell intercalation. Development 2017; 144:2961-2968. [PMID: 28811312 DOI: 10.1242/dev.148940] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 07/05/2017] [Indexed: 01/04/2023]
Abstract
The Drosophila tracheal system consists of an interconnected network of monolayered epithelial tubes that ensures oxygen transport in the larval and adult body. During tracheal dorsal branch (DB) development, individual DBs elongate as a cluster of cells, led by tip cells at the front and trailing cells in the rear. Branch elongation is accompanied by extensive cell intercalation and cell lengthening of the trailing stalk cells. Although cell intercalation is governed by Myosin II (MyoII)-dependent forces during tissue elongation in the Drosophila embryo that lead to germ-band extension, it remained unclear whether MyoII plays a similar active role during tracheal branch elongation and intercalation. Here, we have used a nanobody-based approach to selectively knock down MyoII in tracheal cells. Our data show that, despite the depletion of MyoII function, tip cell migration and stalk cell intercalation (SCI) proceed at a normal rate. This confirms a model in which DB elongation and SCI in the trachea occur as a consequence of tip cell migration, which produces the necessary forces for the branching process.
Collapse
Affiliation(s)
| | - Stefan Harmansa
- Biozentrum, University of Basel, Klingelbergstr. 50/70, 4056 Basel, Switzerland
| | - Emmanuel Caussinus
- Institute of Molecular Life Sciences (IMLS), University of Zurich, 8057 Zurich, Switzerland
| | - Markus Affolter
- Biozentrum, University of Basel, Klingelbergstr. 50/70, 4056 Basel, Switzerland
| |
Collapse
|
32
|
Berndt N, Seib E, Kim S, Troost T, Lyga M, Langenbach J, Haensch S, Kalodimou K, Delidakis C, Klein T. Ubiquitylation-independent activation of Notch signalling by Delta. eLife 2017; 6:27346. [PMID: 28960177 PMCID: PMC5675594 DOI: 10.7554/elife.27346] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 09/28/2017] [Indexed: 12/29/2022] Open
Abstract
Ubiquitylation (ubi) by the E3-ligases Mindbomb1 (Mib1) and Neuralized (Neur) is required for activation of the DSL ligands Delta (Dl) and Serrate (Ser) to activate Notch signalling. These ligases transfer ubiquitin to lysines of the ligands' intracellular domains (ICDs), which sends them into an Epsin-dependent endocytic pathway. Here, we have tested the requirement of ubi of Dl for signalling. We found that Dl requires ubi for its full function, but can also signal in two ubi-independent modes, one dependent and one independent of Neur. We identified two neural lateral specification processes where Dl signals in an ubi-independent manner. Neur, which is needed for these processes, was shown to be able to activate Dl in an ubi-independent manner. Our analysis suggests that one important role of DSL protein ubi by Mib1 is their release from cis-inhibitory interactions with Notch, enabling them to trans-activate Notch on adjacent cells.
Collapse
Affiliation(s)
- Nicole Berndt
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Duesseldorf, Germany
| | - Ekaterina Seib
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Duesseldorf, Germany
| | - Soya Kim
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Duesseldorf, Germany.,Molekulare Zellbiologie, Institut I für Anatomie, Uniklinik Köln, Universität zu Köln, Köln, Germany
| | - Tobias Troost
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Duesseldorf, Germany
| | - Marvin Lyga
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Duesseldorf, Germany
| | - Jessica Langenbach
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Duesseldorf, Germany
| | - Sebastian Haensch
- Center of Advanced Imaging, Heinrich-Heine-Universitaet Duesseldorf, Duesseldorf, Germany
| | - Konstantina Kalodimou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Heraklion, Greece.,Department of Biology, University of Crete, Heraklion, Greece
| | - Christos Delidakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Heraklion, Greece.,Department of Biology, University of Crete, Heraklion, Greece
| | - Thomas Klein
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
33
|
Wortman JC, Nahmad M, Zhang PC, Lander AD, Yu CC. Expanding signaling-molecule wavefront model of cell polarization in the Drosophila wing primordium. PLoS Comput Biol 2017; 13:e1005610. [PMID: 28671940 PMCID: PMC5515495 DOI: 10.1371/journal.pcbi.1005610] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 07/18/2017] [Accepted: 05/26/2017] [Indexed: 01/08/2023] Open
Abstract
In developing tissues, cell polarization and proliferation are regulated by morphogens and signaling pathways. Cells throughout the Drosophila wing primordium typically show subcellular localization of the unconventional myosin Dachs on the distal side of cells (nearest the center of the disc). Dachs localization depends on the spatial distribution of bonds between the protocadherins Fat (Ft) and Dachsous (Ds), which form heterodimers between adjacent cells; and the Golgi kinase Four-jointed (Fj), which affects the binding affinities of Ft and Ds. The Fj concentration forms a linear gradient while the Ds concentration is roughly uniform throughout most of the wing pouch with a steep transition region that propagates from the center to the edge of the pouch during the third larval instar. Although the Fj gradient is an important cue for polarization, it is unclear how the polarization is affected by cell division and the expanding Ds transition region, both of which can alter the distribution of Ft-Ds heterodimers around the cell periphery. We have developed a computational model to address these questions. In our model, the binding affinity of Ft and Ds depends on phosphorylation by Fj. We assume that the asymmetry of the Ft-Ds bond distribution around the cell periphery defines the polarization, with greater asymmetry promoting cell proliferation. Our model predicts that this asymmetry is greatest in the radially-expanding transition region that leaves polarized cells in its wake. These cells naturally retain their bond distribution asymmetry after division by rapidly replenishing Ft-Ds bonds at new cell-cell interfaces. Thus we predict that the distal localization of Dachs in cells throughout the pouch requires the movement of the Ds transition region and the simple presence, rather than any specific spatial pattern, of Fj. In the tissues of a developing organism, specialized proteins can control cell growth and give cells a sense of direction, e.g., which way is the head or the tail, by having their concentration vary throughout the tissue. In cells of the developing fruit fly wing, a protein called Dachs localizes on the side of the cell closest to the center of the tissue, indicating a directionality. The localization of Dachs is determined by the spatial distribution, around the periphery of a cell, of intercellular bonds of the proteins Fat and Dachsous between adjacent cells. Here we asked how this cell directionality is affected when cells divide and when the concentration of Dachsous changes over time. We use a computational model to show that as the circular step-up region of the Dachsous concentration profile sweeps radially outward, like rings radiating outward from where a pebble was dropped in a pond, it leaves polarized cells in its wake. Our model also shows how cells can naturally recover their directionality after cell division.
Collapse
Affiliation(s)
- Juliana C. Wortman
- Department of Physics and Astronomy, University of California, Irvine, Irvine, California, United States of America
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
| | - Marcos Nahmad
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, United States of America
| | - Peng Cheng Zhang
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
- Department of Biomedical Engineering, University of California, Irvine, Irvine, California, United States of America
| | - Arthur D. Lander
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, United States of America
- Department of Biomedical Engineering, University of California, Irvine, Irvine, California, United States of America
| | - Clare C. Yu
- Department of Physics and Astronomy, University of California, Irvine, Irvine, California, United States of America
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
- * E-mail:
| |
Collapse
|
34
|
|
35
|
Belmonte JM, Swat MH, Glazier JA. Filopodial-Tension Model of Convergent-Extension of Tissues. PLoS Comput Biol 2016; 12:e1004952. [PMID: 27322528 PMCID: PMC4913901 DOI: 10.1371/journal.pcbi.1004952] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 04/19/2016] [Indexed: 11/25/2022] Open
Abstract
In convergent-extension (CE), a planar-polarized epithelial tissue elongates (extends) in-plane in one direction while shortening (converging) in the perpendicular in-plane direction, with the cells both elongating and intercalating along the converging axis. CE occurs during the development of most multicellular organisms. Current CE models assume cell or tissue asymmetry, but neglect the preferential filopodial activity along the convergent axis observed in many tissues. We propose a cell-based CE model based on asymmetric filopodial tension forces between cells and investigate how cell-level filopodial interactions drive tissue-level CE. The final tissue geometry depends on the balance between external rounding forces and cell-intercalation traction. Filopodial-tension CE is robust to relatively high levels of planar cell polarity misalignment and to the presence of non-active cells. Addition of a simple mechanical feedback between cells fully rescues and even improves CE of tissues with high levels of polarity misalignments. Our model extends easily to three dimensions, with either one converging and two extending axes, or two converging and one extending axes, producing distinct tissue morphologies, as observed in vivo. The development of an embryo from a fertilized egg to an adult organism requires not only cell proliferation and differentiation, but also numerous types of tissue restructuring. The development of a relatively round initial embryo into one elongated along its rostral-caudal axis involves coordinated tissue elongation and cell reorganization in one or more groups of cells or tissues. Counterintuitively, in many organisms, cells in elongating tissues elongate and increase their protrusive activity in the direction perpendicular to the axis of elongation (convergent extension). Experimental and theoretical studies have not determined how this cell-level oriented protrusive activity leads to observed tissue-level changes in morphology. We propose a filopodial-tension model that shows how tension from oriented cell protrusions leads to observed patterns of tissue CE.
Collapse
Affiliation(s)
- Julio M Belmonte
- Biocomplexity Institute and Department of Physics, Indiana University Bloomington, Bloomington, Indiana, United States of America
| | - Maciej H Swat
- Biocomplexity Institute and Department of Physics, Indiana University Bloomington, Bloomington, Indiana, United States of America
| | - James A Glazier
- Biocomplexity Institute and Department of Intelligent Systems Engineering, Indiana University Bloomington, Bloomington, Indiana, United States of America
| |
Collapse
|
36
|
Abstract
Protein depletion by genetic means, in a very general sense including the use of RNA interference [1, 2] or CRISPR/Cas9-based methods, represents a central paradigm of modern biology to study protein functions in vivo. However, acting upstream the proteic level is a limiting factor if the turnover of the target protein is slow or the existing pool of the target protein is important (for instance, in insect embryos, as a consequence of a strong maternal contribution). In order to circumvent these problems, we developed deGradFP [3, 4]. deGradFP harnesses the ubiquitin-proteasome pathway to achieve direct depletion of GFP-tagged proteins. deGradFP is in essence a universal method because it relies on an evolutionarily conserved machinery for protein catabolism in eukaryotic cells; see refs. 5, 6 for review. deGradFP is particularly convenient in Drosophila melanogaster where it is implemented by a genetically encoded effector expressed under the control of the Gal4 system. deGradFP is a ready-to-use solution to perform knockdowns at the protein level if a fly line carrying a functional GFP-tagged version of the gene of interest is available. Many such lines have already been generated by the Drosophila community through different technologies allowing to make genomic rescue constructs or direct GFP knockins: protein-trap stock collections [7, 8] ( http://cooley.medicine.yale.edu/flytrap/ , http://www.flyprot.org/ ), P[acman] system [9], MiMIC lines [10, 11], and CRISPR/Cas9-driven homologous recombination.Two essential controls of a protein knockdown experiment are easily achieved using deGradFP. First, the removal of the target protein can be assessed by monitoring the disappearance of the GFP tag by fluorescence microscopy in parallel to the documentation of the phenotype of the protein knockdown (see Note 1 ). Second, the potential nonspecific effects of deGradFP can be assessed in control fly lacking a GFP-tagged target protein. So far, no nonspecific effects of the deGradFP effector have been reported [3].
Collapse
Affiliation(s)
- Emmanuel Caussinus
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Markus Affolter
- Growth & Development, Biozentrum, University of Basel, Room 200B, Klingelbergstrasse 50/70, 4056, Basel, Switzerland.
| |
Collapse
|
37
|
|
38
|
Dpp spreading is required for medial but not for lateral wing disc growth. Nature 2015; 527:317-22. [PMID: 26550827 DOI: 10.1038/nature15712] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 09/10/2015] [Indexed: 01/16/2023]
Abstract
Drosophila Decapentaplegic (Dpp) has served as a paradigm to study morphogen-dependent growth control. However, the role of a Dpp gradient in tissue growth remains highly controversial. Two fundamentally different models have been proposed: the 'temporal rule' model suggests that all cells of the wing imaginal disc divide upon a 50% increase in Dpp signalling, whereas the 'growth equalization model' suggests that Dpp is only essential for proliferation control of the central cells. Here, to discriminate between these two models, we generated and used morphotrap, a membrane-tethered anti-green fluorescent protein (GFP) nanobody, which enables immobilization of enhanced (e)GFP::Dpp on the cell surface, thereby abolishing Dpp gradient formation. We find that in the absence of Dpp spreading, wing disc patterning is lost; however, lateral cells still divide at normal rates. These data are consistent with the growth equalization model, but do not fit a global temporal rule model in the wing imaginal disc.
Collapse
|
39
|
Hariharan IK. Organ Size Control: Lessons from Drosophila. Dev Cell 2015; 34:255-65. [PMID: 26267393 DOI: 10.1016/j.devcel.2015.07.012] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 07/13/2015] [Accepted: 07/22/2015] [Indexed: 12/23/2022]
Abstract
Of fundamental interest to biologists is how organs achieve a reproducible size during development. Studies of the developing Drosophila wing have provided many key insights that will help give a conceptual understanding of the process beyond the fly. In the wing, there is evidence for both "top-down" mechanisms, in which signals emanating from small subsets of cells direct global proliferation, and "bottom-up" mechanisms, in which the final size is an emergent property of local cell-cell interactions. Mechanical forces also appear to have an important role along with the Hippo pathway, which may integrate multiple types of inputs to regulate the extent of growth.
Collapse
Affiliation(s)
- Iswar K Hariharan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
40
|
The transcription factor optomotor-blind antagonizes Drosophila haltere growth by repressing decapentaplegic and hedgehog targets. PLoS One 2015; 10:e0121239. [PMID: 25793870 PMCID: PMC4368094 DOI: 10.1371/journal.pone.0121239] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 01/29/2015] [Indexed: 12/23/2022] Open
Abstract
In Drosophila, decapentaplegic, which codes for a secreted signaling molecule, is activated by the Hedgehog signaling pathway at the anteroposterior compartment border of the two dorsal primordia; the wing and the haltere imaginal discs. In the wing disc, Decapentaplegic and Hedgehog signaling targets are implicated in cell proliferation and cell survival. However, most of their known targets in the wing disc are not expressed in the haltere disc due to their repression by the Hox gene Ultrabithorax. The T-box gene optomotor-blind escapes this repression in the haltere disc, and therefore is expressed in both the haltere and wing discs. Optomotor-blind is a major player during wing development and its function has been intensely investigated in this tissue, however, its role in haltere development has not been reported so far. Here we show that Optomotor-blind function in the haltere disc differs from that in the wing disc. Unlike its role in the wing, Optomotor-blind does not prevent apoptosis in the haltere but rather limits growth by repressing several Decapentaplegic and Hedgehog targets involved both in wing proliferation and in modulating the spread of morphogens similar to Ultrabithorax function but without disturbing Ultrabithorax expression.
Collapse
|
41
|
Ducuing A, Keeley C, Mollereau B, Vincent S. A DPP-mediated feed-forward loop canalizes morphogenesis during Drosophila dorsal closure. ACTA ACUST UNITED AC 2015; 208:239-48. [PMID: 25601405 PMCID: PMC4298692 DOI: 10.1083/jcb.201410042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
During Drosophila dorsal closure, DPP and JNK signaling form a feed-forward loop that controls the specification and differentiation of leading edge cells to ensure robust morphogenesis. Development is robust because nature has selected various mechanisms to buffer the deleterious effects of environmental and genetic variations to deliver phenotypic stability. Robustness relies on smart network motifs such as feed-forward loops (FFLs) that ensure the reliable interpretation of developmental signals. In this paper, we show that Decapentaplegic (DPP) and JNK form a coherent FFL that controls the specification and differentiation of leading edge cells during Drosophila melanogaster dorsal closure (DC). We provide molecular evidence that through repression by Brinker (Brk), the DPP branch of the FFL filters unwanted JNK activity. High-throughput live imaging revealed that this DPP/Brk branch is dispensable for DC under normal conditions but is required when embryos are subjected to thermal stress. Our results indicate that the wiring of DPP signaling buffers against environmental challenges and canalizes cell identity. We propose that the main function of DPP pathway during Drosophila DC is to ensure robust morphogenesis, a distinct function from its well-established ability to spread spatial information.
Collapse
Affiliation(s)
- Antoine Ducuing
- Laboratory of Molecular Biology of the Cell, UMR5239, Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique, 69007 Lyon, France
| | - Charlotte Keeley
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Bertrand Mollereau
- Laboratory of Molecular Biology of the Cell, UMR5239, Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique, 69007 Lyon, France
| | - Stéphane Vincent
- Laboratory of Molecular Biology of the Cell, UMR5239, Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique, 69007 Lyon, France
| |
Collapse
|
42
|
Heemskerk I, Lecuit T, LeGoff L. Dynamic clonal analysis based on chronic in vivo imaging allows multiscale quantification of growth in the Drosophila wing disc. Development 2014; 141:2339-48. [PMID: 24866118 DOI: 10.1242/dev.109264] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In the course of morphogenesis, tissues change shape and grow. How this is orchestrated is largely unknown, partly owing to the lack of experimental methods to visualize and quantify growth. Here, we describe a novel experimental approach to investigate the growth of tissues in vivo on a time-scale of days, as employed to study the Drosophila larval imaginal wing disc, the precursor of the adult wing. We developed a protocol to image wing discs at regular intervals in living anesthetized larvae so as to follow the growth of the tissue over extended periods of time. This approach can be used to image cells at high resolution in vivo. At intermediate scale, we tracked the increase in cell number within clones as well as the changes in clone area and shape. At scales extending to the tissue level, clones can be used as landmarks for measuring strain, as a proxy for growth. We developed general computational tools to extract strain maps from clonal shapes and landmark displacements in individual tissues, and to combine multiple datasets into a mean strain. In the disc, we use these to compare properties of growth at the scale of clones (a few cells) and at larger regional scales.
Collapse
Affiliation(s)
- Idse Heemskerk
- Kavli Institute for Theoretical Physics, University of California, Santa Barbara, CA 93106, USA
| | - Thomas Lecuit
- Aix Marseille Université, CNRS, IBDML UMR7288, case 907, Marseille 13009, France
| | - Loïc LeGoff
- Aix Marseille Université, CNRS, IBDML UMR7288, case 907, Marseille 13009, France
| |
Collapse
|
43
|
Vincent JP. Modulating and measuring Wingless signalling. Methods 2014; 68:194-8. [PMID: 24675402 DOI: 10.1016/j.ymeth.2014.03.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 03/10/2014] [Accepted: 03/13/2014] [Indexed: 12/19/2022] Open
Abstract
The main Wnt ligand of Drosophila activates a conserved canonical signalling pathway to regulate a plethora of cellular activities during development, regeneration and nervous system function. Here I first describe experimental means of measuring and modulating Wingless signalling in Drosophila cell culture. Various reporters have been devised by placing TCF-binding sites or DNA fragments from known target genes upstream of luciferase-coding sequences. Signalling can be activated in cells by addition of Wingless conditioned medium, treatment with a chemical inhibitor of Shaggy/GSK3 or transfection with a plasmid encoding activated Armadillo (Drosophila β-catenin). Measuring Wingless signalling in intact tissue is somewhat more challenging than in cell culture. Synthetic transgenic reporters have been devised but further improvements are needed to achieve sensitive responsiveness to Wingless at all times and places. As an alternative, gene traps in frizzled3 and notum/wingful, two context-independent endogenous targets, can be used as reporters. It is hoped that further modification of these loci could lead to more versatile and sensitive means of detecting signalling. Many genetic tools are available to trigger ectopic signalling or prevent endogenous signalling. These mostly rely on RNAi-producing transgenes or the generation of mutant patches by mitotic recombination. New developments in genome engineering are opening further means of manipulating the components of Wingless signalling with exquisite temporal and spatial precision.
Collapse
Affiliation(s)
- Jean-Paul Vincent
- MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW71AA, United Kingdom.
| |
Collapse
|
44
|
Affiliation(s)
- Ginés Morata
- Centro de Biología Molecular, CSIC-UAM, Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Gary Struhl
- Department of Genetics and Development, Columbia University, New York, New York 10032, USA
| |
Collapse
|
45
|
LeGoff L, Rouault H, Lecuit T. A global pattern of mechanical stress polarizes cell divisions and cell shape in the growing Drosophila wing disc. Development 2013; 140:4051-9. [DOI: 10.1242/dev.090878] [Citation(s) in RCA: 194] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Organismal development is under genetic control. Ultimately, mechanical forces shape embryos. If we want to understand the precise regulation of size and shape in animals, we must dissect how forces are distributed in developing tissues, and how they drive cell behavior to shape organs. This has not been addressed fully in the context of growing tissues. As cells grow and divide, they exert a pressure on their neighbors. How these local stresses add up or dissipate as the tissue grows is an unanswered question. We address this issue in the growing wing imaginal disc of Drosophila larvae, the precursor of the adult wing. We used a quantitative approach to analyze the strains and stresses of cells of the wing pouch, and found a global pattern of stress whereby cells in the periphery of the tissue are mechanically stretched and cells in the center are compressed. This pattern has important consequences on cell shape in the wing pouch: cells respond to it by polarizing their acto-myosin cortex, and aligning their divisions with the main axis of cell stretch, thereby polarizing tissue growth. Ectopic perturbations of tissue growth by the Hippo signaling pathway reorganize this pattern in a non-autonomous manner, suggesting a synergy between tissue mechanics and growth control during wing disc morphogenesis.
Collapse
Affiliation(s)
- Loïc LeGoff
- IBDML, UMR7288 CNRS-Université d’Aix-Marseille. Campus de Luminy, case 907, 13288 Marseille Cedex 09, France
| | - Hervé Rouault
- IBDML, UMR7288 CNRS-Université d’Aix-Marseille. Campus de Luminy, case 907, 13288 Marseille Cedex 09, France
| | - Thomas Lecuit
- IBDML, UMR7288 CNRS-Université d’Aix-Marseille. Campus de Luminy, case 907, 13288 Marseille Cedex 09, France
| |
Collapse
|
46
|
Ayala-Camargo A, Anderson AM, Amoyel M, Rodrigues AB, Flaherty MS, Bach EA. JAK/STAT signaling is required for hinge growth and patterning in the Drosophila wing disc. Dev Biol 2013; 382:413-26. [PMID: 23978534 DOI: 10.1016/j.ydbio.2013.08.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 07/31/2013] [Accepted: 08/18/2013] [Indexed: 01/15/2023]
Abstract
JAK/STAT signaling is localized to the wing hinge, but its function there is not known. Here we show that the Drosophila STAT Stat92E is downstream of Homothorax and is required for hinge development by cell-autonomously regulating hinge-specific factors. Within the hinge, Stat92E activity becomes restricted to gap domain cells that lack Nubbin and Teashirt. While gap domain cells lacking Stat92E have significantly reduced proliferation, increased JAK/STAT signaling there does not expand this domain. Thus, this pathway is necessary but not sufficient for gap domain growth. We show that reduced Wingless (Wg) signaling dominantly inhibits Stat92E activity in the hinge. However, ectopic JAK/STAT signaling does not perturb Wg expression in the hinge. We report negative interactions between Stat92E and the notum factor Araucan, resulting in restriction of JAK/STAT signaling from the notum. In addition, we find that the distal factor Nub represses the ligand unpaired as well as Stat92E activity. These data suggest that distal expansion of JAK/STAT signaling is deleterious to wing blade development. Indeed, mis-expression of Unpaired within the presumptive wing blade causes small, stunted adult wings. We conclude that JAK/STAT signaling is critical for hinge fate specification and growth of the gap domain and that its restriction to the hinge is required for proper wing development.
Collapse
Affiliation(s)
- Aidee Ayala-Camargo
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York 10016-6402, USA
| | | | | | | | | | | |
Collapse
|
47
|
Perea D, Molohon K, Edwards K, Díaz-Benjumea FJ. Multiple roles of the gene zinc finger homeodomain-2 in the development of the Drosophila wing. Mech Dev 2013; 130:467-81. [PMID: 23811114 DOI: 10.1016/j.mod.2013.06.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/05/2013] [Accepted: 06/11/2013] [Indexed: 10/26/2022]
Abstract
The gene zfh2 and its human homolog Atbf1 encode huge molecules with several homeo- and zinc finger domains. It has been reported that they play important roles in neural differentiation and promotion of apoptosis in several tissues of both humans and flies. In the Drosophila wing imaginal disc, Zfh2 is expressed in a dynamic pattern and previous results suggest that it is involved is proximal-distal patterning. In this report we go further in the analysis of the function of this gene in wing development, performing ectopic expression experiments and studying its effects in genes involved in wing development. Our results suggest that Zfh2 plays an important role controlling the expression of several wing genes and in the specification of those cellular properties that define the differences in cell proliferation between proximal and distal domains of the wing disc.
Collapse
Affiliation(s)
- Daniel Perea
- Centro de Biología Molecular-Severo Ochoa, Universidad Autónoma-Cantoblanco, 28049 Madrid, Spain
| | | | | | | |
Collapse
|
48
|
Shaping organs by a wingless-int/Notch/nonmuscle myosin module which orients feather bud elongation. Proc Natl Acad Sci U S A 2013; 110:E1452-61. [PMID: 23576731 DOI: 10.1073/pnas.1219813110] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
How organs are shaped to specific forms is a fundamental issue in developmental biology. To address this question, we used the repetitive, periodic pattern of feather morphogenesis on chicken skin as a model. Avian feathers within a single tract extend from dome-shaped primordia to thin conical structures with a common axis of orientation. From a systems biology perspective, the process is precise and robust. Using tissue transplantation assays, we demonstrate that a "zone of polarizing activity," localized in the posterior feather bud, is necessary and sufficient to mediate the directional elongation. This region contains a spatially well-defined nuclear β-catenin zone, which is induced by wingless-int (Wnt)7a protein diffusing in from posterior bud epithelium. Misexpressing nuclear β-catenin randomizes feather polarity. This dermal nuclear β-catenin zone, surrounded by Notch1 positive dermal cells, induces Jagged1. Inhibition of Notch signaling disrupts the spatial configuration of the nuclear β-catenin zone and leads to randomized feather polarity. Mathematical modeling predicts that lateral inhibition, mediated by Notch signaling, functions to reduce Wnt7a gradient variations and fluctuations to form the sharp boundary observed for the dermal β-catenin zone. This zone is also enriched for nonmuscle myosin IIB. Suppressing nonmuscle myosin IIB disrupts directional cell rearrangements and abolishes feather bud elongation. These data suggest that a unique molecular module involving chemical-mechanical coupling converts a pliable chemical gradient to a precise domain, ready for subsequent mechanical action, thus defining the position, boundary, and duration of localized morphogenetic activity that molds the shape of growing organs.
Collapse
|
49
|
Dpp-induced Egfr signaling triggers postembryonic wing development in Drosophila. Proc Natl Acad Sci U S A 2013; 110:5058-63. [PMID: 23479629 DOI: 10.1073/pnas.1217538110] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The acquisition of flight contributed to the success of insects and winged forms are present in most orders. Key to understanding the origin of wings will be knowledge of the earliest postembryonic events promoting wing outgrowth. The Drosophila melanogaster wing is intensely studied as a model appendage, and yet little is known about the beginning of wing outgrowth. Vein (Vn) is a neuregulin-like ligand for the EGF receptor (Egfr), which is necessary for global development of the early Drosophila wing disc. vn is not expressed in the embryonic wing primordium and thus has to be induced de novo in the nascent larval wing disc. We find that Decapentaplegic (Dpp), a Bone Morphogenetic Protein (BMP) family member, provides the instructive signal for initiating vn expression. The signaling involves paracrine communication between two epithelia in the early disc. Once initiated, vn expression is amplified and maintained by autocrine signaling mediated by the E-twenty six (ETS)-factor PointedP2 (PntP2). This interplay of paracrine and autocrine signaling underlies the spatial and temporal pattern of induction of Vn/Egfr target genes and explains both body wall development and wing outgrowth. It is possible this gene regulatory network governing expression of an EGF ligand is conserved and reflects a common origin of insect wings.
Collapse
|
50
|
Hatini V, Kula-Eversole E, Nusinow D, Del Signore SJ. Essential roles for stat92E in expanding and patterning the proximodistal axis of the Drosophila wing imaginal disc. Dev Biol 2013; 378:38-50. [PMID: 23499656 DOI: 10.1016/j.ydbio.2013.02.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 02/17/2013] [Accepted: 02/20/2013] [Indexed: 12/26/2022]
Abstract
The Drosophila wing imaginal disc is subdivided along the proximodistal axis into the distal pouch, the hinge, the surrounding pleura, and the notum. While the genetic pathways that specify the identity of each of these domains have been well studied, the mechanisms that coordinate the relative expansion of these domains are not well understood. Here we investigated the role of the stat92E signal transducer and activator of transcription in wing proximodistal development. We find that stat92E is active ubiquitously in early wing imaginal discs, where it acts to inhibit the induction of ectopic wing fields. Subsequently, stat92E activity is down regulated in the notum and distal pouch. These dynamics coincide with and contribute to the proportional subdivision and expansion of these primordia. As development proceeds, stat92E activity becomes restricted to the hinge, where it promotes normal expansion of the hinge, and restricts expansion of the notum. We also find that stat92E is required autonomously to specify dorsal pleura identity and inhibit notum identity to properly subdivide the body wall. Our data suggest that stat92E activity is regulated along the proximodistal axis to pattern this axis and control the relative expansion of the pouch, hinge, and notum.
Collapse
Affiliation(s)
- Victor Hatini
- Tufts University School of Medicine, Department of Anatomy & Cellular Biology, Program in Cell, Molecular and Developmental Biology, 150 Harrison Avenue, Boston, MA 02111, USA.
| | | | | | | |
Collapse
|